1
|
Li X, Fu B, Zhao C, Hu J, Zhang X, Fu Y, She X, Gu C, Cheng M, Wang F, Song X, Dai J, Yin J, Fu Y, Zheng P, Wu F, Zhu Y, Ma K, Gao X, Wang M, Zeng Q, Cui B. Early-life noise exposure causes cognitive impairment in a sex-dependent manner by disrupting homeostasis of the microbiota-gut-brain axis. Brain Behav Immun 2023; 114:221-239. [PMID: 37648006 DOI: 10.1016/j.bbi.2023.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/01/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
Epidemiological investigations show that noise exposure in early life is associated with health and cognitive impairment. The gut microbiome established in early life plays a crucial role in modulating developmental processes that subsequently affect brain function and behavior. Here, we examined the impact of early-life exposure to noise on cognitive function in adolescent rats by analyzing the gut microbiome and metabolome to elucidate the underlying mechanisms. Chronic noise exposure during early life led to cognitive deficits, hippocampal injury, and neuroinflammation. Early-life noise exposure showed significant difference on the composition and function of the gut microbiome throughout adolescence, subsequently causing axis-series changes in fecal short-chain fatty acid (SCFA) metabolism and serum metabolome profiles, as well as dysregulation of endothelial tight junction proteins, in both intestine and brain. We also observed sex-dependent effects of microbiota depletion on SCFA-related beneficial bacteria in adolescence. Experiments on microbiota transplantation and SCFA supplementation further confirmed the role of intestinal bacteria and related SCFAs in early-life noise-exposure-induced impairments in cognition, epithelial integrity, and neuroinflammation. Overall, these results highlight the homeostatic imbalance of microbiota-gut-brain axis as an important physiological response toward environmental noise during early life and reveals subtle differences in molecular signaling processes between male and female rats.
Collapse
Affiliation(s)
- Xiaofang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Public Health and Management, Binzhou Medical University, Yantai 264003, China
| | - Bo Fu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Chunli Zhao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Public Health and Management, Binzhou Medical University, Yantai 264003, China
| | - Junjie Hu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xinyao Zhang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yiming Fu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Cui Gu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Mengzhu Cheng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Fenghan Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xiaoqiong Song
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jie Dai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jiayi Yin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yu Fu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Pengfang Zheng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Fangshan Wu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yingwen Zhu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Kefeng Ma
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Miao Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Qiang Zeng
- Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Public Health and Management, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
2
|
Zhuang H, Li Q, Sun C, Xu D, Gan G, Zhang C, Chen C, Yuan Y, Liu L, Xiao Y, Yao X, Wang C, Kang X, Yang C, Zhao J, Chen W, Wang J, Li J, Luo C, Wang J, Jia X, Yu Z, Liu L. Voluntary wheel exercise ameliorates cognitive impairment, hippocampal neurodegeneration and microglial abnormalities preceded by demyelination in a male mouse model of noise-induced hearing loss. Brain Behav Immun 2023; 114:325-348. [PMID: 37683962 DOI: 10.1016/j.bbi.2023.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Acquired peripheral hearing loss (APHL) in midlife has been identified as the greatest modifiable risk factor for dementia; however, the pathophysiological neural mechanisms linking APHL with an increased risk of dementia remain to be elucidated. Here, in an adult male mouse model of noise-induced hearing loss (NIHL), one of the most common forms of APHL, we demonstrated accelerated age-related cognitive decline and hippocampal neurodegeneration during a 6-month follow-up period, accompanied by progressive hippocampal microglial aberrations preceded by immediate-onset transient elevation in serum glucocorticoids and delayed-onset sustained myelin disruption in the hippocampus. Pretreatment with the glucocorticoid receptor antagonist RU486 before stressful noise exposure partially mitigated the early activation of hippocampal microglia, which were present at 7 days post noise exposure (7DPN), but had no impact on later microglial aberrations, hippocampal neurodegeneration, or cognitive decline exhibited at 1 month post noise exposure (1MPN). One month of voluntary wheel exercise following noise exposure barely affected either the hearing threshold shift or hippocampal myelin changes but effectively countered cognitive impairment and the decline in hippocampal neurogenesis in NIHL mice at 1MPN, paralleled by the normalization of microglial morphology, which coincided with a reduction in microglial myelin inclusions and a restoration of microglial hypoxia-inducible factor-1α (HIF1α) expression. Our results indicated that accelerated cognitive deterioration and hippocampal neuroplastic decline following NIHL are most likely driven by the maladaptive response of hippocampal microglia to myelin damage secondary to hearing loss, and we also demonstrated the potential of voluntary physical exercise as a promising and cost-effective strategy to alleviate the detrimental impact of APHL on cognitive function and thus curtail the high and continuously increasing global burden of dementia. Furthermore, the findings of the present study highlight the contribution of myelin debris overload to microglial malfunction and identify the microglial HIF1α-related pathway as an attractive candidate for future comprehensive investigation to obtain a more definitive picture of the underlying mechanisms linking APHL and dementia.
Collapse
Affiliation(s)
- Hong Zhuang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qian Li
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Congli Sun
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Dan Xu
- School of Public Health, Southeast University, Nanjing 210009, China
| | - Guangming Gan
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchen Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chen Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Yang Yuan
- Department of Endocrinology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China
| | - Linchen Liu
- Department of Rheumatology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Xiao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiuting Yao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Conghui Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaoming Kang
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Chenxi Yang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jingyi Zhao
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Wenhao Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jiatang Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jinyu Li
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Caichen Luo
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Xirui Jia
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Zhehao Yu
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
3
|
Zhou X, Jin L, Li Y, Wang Y, Li W, Shen X. Comprehensive analysis of N6-methyladenosine-related RNA methylation in the mouse hippocampus after acquired hearing loss. BMC Genomics 2023; 24:577. [PMID: 37759187 PMCID: PMC10537436 DOI: 10.1186/s12864-023-09697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The mechanism underlying cognitive impairment after hearing loss (HL) remains unclear. N6-methyladenosine (m6A) is involved in many neurodegenerative diseases; however, its role in cognitive impairment after HL has not yet been investigated. Therefore, we aimed to analyze the m6A modification profile of the mouse hippocampus after HL exposure. A mouse model of neomycin-induced HL was established. An auditory brainstem-response test was utilized for detecting hearing threshold. The passive avoidance test was served as the mean for evaluating cognitive function. The m6A-regulated enzyme expression levels were analyzed by using reverse transcription quantitative real-time polymerase chain reaction and western blot analyses. RNA sequencing (RNA-Seq) and methylated RNA immunoprecipitation sequencing (MeRIP-Seq) were performed with the aim of investigating gene expression differences and m6A modification in the mouse hippocampus. RESULTS Neomycin administration induced severe HL in mice. At four months of age, the mice in the HL group showed poorer cognitive performance than the mice in the control group. METTL14, WTAP, and YTHDF2 mRNA levels were downregulated in the hippocampi of HL mice, whereas ALKBH5 and FTO mRNA levels were significantly upregulated. At the protein level, METTL3 and FTO were significantly upregulated. Methylated RNA immunoprecipitation sequencing analysis revealed 387 and 361 m6A hypermethylation and hypomethylation peaks, respectively. Moreover, combined analysis of mRNA expression levels and m6A peaks revealed eight mRNAs with significantly changed expression levels and methylation. CONCLUSIONS Our findings revealed the m6A transcriptome-wide profile in the hippocampus of HL mice, which may provide a basis for understanding the association between HL and cognitive impairment from the perspective of epigenetic modifications.
Collapse
Affiliation(s)
- Xuehua Zhou
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Lin Jin
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Yufeng Li
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Yiru Wang
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Wen Li
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Xia Shen
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China.
| |
Collapse
|
4
|
Pisani A, Paciello F, Del Vecchio V, Malesci R, De Corso E, Cantone E, Fetoni AR. The Role of BDNF as a Biomarker in Cognitive and Sensory Neurodegeneration. J Pers Med 2023; 13:jpm13040652. [PMID: 37109038 PMCID: PMC10140880 DOI: 10.3390/jpm13040652] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has a crucial function in the central nervous system and in sensory structures including olfactory and auditory systems. Many studies have highlighted the protective effects of BDNF in the brain, showing how it can promote neuronal growth and survival and modulate synaptic plasticity. On the other hand, conflicting data about BDNF expression and functions in the cochlear and in olfactory structures have been reported. Several clinical and experimental research studies showed alterations in BDNF levels in neurodegenerative diseases affecting the central and peripheral nervous system, suggesting that BDNF can be a promising biomarker in most neurodegenerative conditions, including Alzheimer's disease, shearing loss, or olfactory impairment. Here, we summarize current research concerning BDNF functions in brain and in sensory domains (olfaction and hearing), focusing on the effects of the BDNF/TrkB signalling pathway activation in both physiological and pathological conditions. Finally, we review significant studies highlighting the possibility to target BDNF as a biomarker in early diagnosis of sensory and cognitive neurodegeneration, opening new opportunities to develop effective therapeutic strategies aimed to counteract neurodegeneration.
Collapse
Affiliation(s)
- Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Del Vecchio
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Malesci
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Eugenio De Corso
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elena Cantone
- Department of Neuroscience, Reproductive Sciences and Dentistry-ENT Section, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
5
|
Paciello F, Ripoli C, Fetoni AR, Grassi C. Redox Imbalance as a Common Pathogenic Factor Linking Hearing Loss and Cognitive Decline. Antioxidants (Basel) 2023; 12:antiox12020332. [PMID: 36829891 PMCID: PMC9952092 DOI: 10.3390/antiox12020332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Experimental and clinical data suggest a tight link between hearing and cognitive functions under both physiological and pathological conditions. Indeed, hearing perception requires high-level cognitive processes, and its alterations have been considered a risk factor for cognitive decline. Thus, identifying common pathogenic determinants of hearing loss and neurodegenerative disease is challenging. Here, we focused on redox status imbalance as a possible common pathological mechanism linking hearing and cognitive dysfunctions. Oxidative stress plays a critical role in cochlear damage occurring during aging, as well as in that induced by exogenous factors, including noise. At the same time, increased oxidative stress in medio-temporal brain regions, including the hippocampus, is a hallmark of neurodegenerative disorders like Alzheimer's disease. As such, antioxidant therapy seems to be a promising approach to prevent and/or counteract both sensory and cognitive neurodegeneration. Here, we review experimental evidence suggesting that redox imbalance is a key pathogenetic factor underlying the association between sensorineural hearing loss and neurodegenerative diseases. A greater understanding of the pathophysiological mechanisms shared by these two diseased conditions will hopefully provide relevant information to develop innovative and effective therapeutic strategies.
Collapse
Affiliation(s)
- Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-0630154966
| | - Anna Rita Fetoni
- Unit of Audiology, Department of Neuroscience, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
6
|
Johne M, Helgers SOA, Alam M, Jelinek J, Hubka P, Krauss JK, Scheper V, Kral A, Schwabe K. Processing of auditory information in forebrain regions after hearing loss in adulthood: Behavioral and electrophysiological studies in a rat model. Front Neurosci 2022; 16:966568. [DOI: 10.3389/fnins.2022.966568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/20/2022] [Indexed: 11/12/2022] Open
Abstract
BackgroundHearing loss was proposed as a factor affecting development of cognitive impairment in elderly. Deficits cannot be explained primarily by dysfunctional neuronal networks within the central auditory system. We here tested the impact of hearing loss in adult rats on motor, social, and cognitive function. Furthermore, potential changes in the neuronal activity in the medial prefrontal cortex (mPFC) and the inferior colliculus (IC) were evaluated.Materials and methodsIn adult male Sprague Dawley rats hearing loss was induced under general anesthesia with intracochlear injection of neomycin. Sham-operated and naive rats served as controls. Postsurgical acoustically evoked auditory brainstem response (ABR)-measurements verified hearing loss after intracochlear neomycin-injection, respectively, intact hearing in sham-operated and naive controls. In intervals of 8 weeks and up to 12 months after surgery rats were tested for locomotor activity (open field) and coordination (Rotarod), for social interaction and preference, and for learning and memory (4-arms baited 8-arms radial maze test). In a final setting, electrophysiological recordings were performed in the mPFC and the IC.ResultsLocomotor activity did not differ between deaf and control rats, whereas motor coordination on the Rotarod was disturbed in deaf rats (P < 0.05). Learning the concept of the radial maze test was initially disturbed in deaf rats (P < 0.05), whereas retesting every 8 weeks did not show long-term memory deficits. Social interaction and preference was also not affected by hearing loss. Final electrophysiological recordings in anesthetized rats revealed reduced firing rates, enhanced irregular firing, and reduced oscillatory theta band activity (4–8 Hz) in the mPFC of deaf rats as compared to controls (P < 0.05). In the IC, reduced oscillatory theta (4–8 Hz) and gamma (30–100 Hz) band activity was found in deaf rats (P < 0.05).ConclusionMinor and transient behavioral deficits do not confirm direct impact of long-term hearing loss on cognitive function in rats. However, the altered neuronal activities in the mPFC and IC after hearing loss indicate effects on neuronal networks in and outside the central auditory system with potential consequences on cognitive function.
Collapse
|
7
|
Patel SV, DeCarlo CM, Book SA, Schormans AL, Whitehead SN, Allman BL, Hayes SH. Noise exposure in early adulthood causes age-dependent and brain region-specific impairments in cognitive function. Front Neurosci 2022; 16:1001686. [PMID: 36312027 PMCID: PMC9606802 DOI: 10.3389/fnins.2022.1001686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Hearing loss is a chronic health condition that affects millions of people worldwide. In addition to age-related hearing impairment, excessive noise exposure is a leading cause of hearing loss. Beyond the devastating effects of hearing impairment itself, epidemiological studies have identified hearing loss as a major risk factor for age-related cognitive decline, including dementia. At present, we currently lack a full understanding of the brain regions and underlying molecular changes that are responsible for mediating the link between hearing loss and cognitive impairment across aging. In the present study, we exposed 6-month-old rats to an occupational-like noise (100 dB SPL, 4 h/day × 30 days) or sham exposure and investigated both hippocampal-dependent (i.e., spatial learning and memory, assessed using the Morris water maze) and striatal-dependent (i.e., visuomotor associative learning, assessed using an operant-conditioning task) cognitive function across aging at 7, 10, and 13 months of age. We also investigated brain region-specific changes in microglial expression following noise/sham exposure in order to assess the potential contribution of this cell type to noise-induced cognitive impairments. Consistent with human studies, the occupational-like noise exposure resulted in high-frequency hearing loss, evidenced by a significant increase in hearing thresholds at 20 kHz. Ultimately, our results suggest that not all higher-level cognitive tasks or their associated brain regions appear to be equally susceptible to noise-induced deficits during aging, as the occupational-like noise exposure caused an age-dependent deficit in spatial but not visuomotor associative learning, as well as altered microglial expression in the hippocampus but not the striatum. Interestingly, we found no significant relationships between spatial learning ability and the level of hearing loss or altered microglial density in the hippocampus following noise exposure, suggesting that other changes in the brain likely contribute to hippocampal-dependent cognitive dysfunction following noise exposure. Lastly, we found that a subset of younger animals also showed noise-induced deficits in spatial learning; findings which suggest that noise exposure may represent an increased risk for cognitive impairment in vulnerable subjects. Overall, our findings highlight that even a mild occupational-like noise exposure earlier in adulthood can have long lasting implications for cognitive function later in life.
Collapse
|
8
|
Manohar S, Chen GD, Ding D, Liu L, Wang J, Chen YC, Chen L, Salvi R. Unexpected Consequences of Noise-Induced Hearing Loss: Impaired Hippocampal Neurogenesis, Memory, and Stress. Front Integr Neurosci 2022; 16:871223. [PMID: 35619926 PMCID: PMC9127992 DOI: 10.3389/fnint.2022.871223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
Noise-induced hearing loss (NIHL), caused by direct damage to the cochlea, reduces the flow of auditory information to the central nervous system, depriving higher order structures, such as the hippocampus with vital sensory information needed to carry out complex, higher order functions. Although the hippocampus lies outside the classical auditory pathway, it nevertheless receives acoustic information that influence its activity. Here we review recent results that illustrate how NIHL and other types of cochlear hearing loss disrupt hippocampal function. The hippocampus, which continues to generate new neurons (neurogenesis) in adulthood, plays an important role in spatial navigation, memory, and emotion. The hippocampus, which contains place cells that respond when a subject enters a specific location in the environment, integrates information from multiple sensory systems, including the auditory system, to develop cognitive spatial maps to aid in navigation. Acute exposure to intense noise disrupts the place-specific firing patterns of hippocampal neurons, "spatially disorienting" the cells for days. More traumatic sound exposures that result in permanent NIHL chronically suppresses cell proliferation and neurogenesis in the hippocampus; these structural changes are associated with long-term spatial memory deficits. Hippocampal neurons, which contain numerous glucocorticoid hormone receptors, are part of a complex feedback network connected to the hypothalamic-pituitary (HPA) axis. Chronic exposure to intense intermittent noise results in prolonged stress which can cause a persistent increase in corticosterone, a rodent stress hormone known to suppress neurogenesis. In contrast, a single intense noise exposure sufficient to cause permanent hearing loss produces only a transient increase in corticosterone hormone. Although basal corticosterone levels return to normal after the noise exposure, glucocorticoid receptors (GRs) in the hippocampus remain chronically elevated. Thus, NIHL disrupts negative feedback from the hippocampus to the HPA axis which regulates the release of corticosterone. Preclinical studies suggest that the noise-induced changes in hippocampal place cells, neurogenesis, spatial memory, and glucocorticoid receptors may be ameliorated by therapeutic interventions that reduce oxidative stress and inflammation. These experimental results may provide new insights on why hearing loss is a risk factor for cognitive decline and suggest methods for preventing this decline.
Collapse
Affiliation(s)
- Senthilvelan Manohar
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Guang-Di Chen
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Lijie Liu
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| | - Jian Wang
- School of Communication Science and Disorders, Dalhousie University, Halifax, NS, Canada
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lin Chen
- Auditory Research Laboratory, University of Science and Technology of China, Hefei, China
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
9
|
Alinaghipour A, Ashabi G, Riahi E, Soheili M, Salami M, Nabavizadeh F. Effects of nano-curcumin on noise stress-induced hippocampus-dependent memory impairment: behavioral and electrophysiological aspects. Pharmacol Rep 2022; 74:461-469. [DOI: 10.1007/s43440-022-00354-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/11/2022]
|
10
|
Parker A, Hobson L, Bains R, Wells S, Bowl M. Investigating audible and ultrasonic noise in modern animal facilities. F1000Res 2022; 11:651. [PMID: 35949916 PMCID: PMC9334837 DOI: 10.12688/f1000research.111170.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
Background: The environmental housing conditions of laboratory animals are important for both welfare and reliable, reproducible data. Guidelines currently exist for factors such as lighting cycles, temperature, humidity, and noise, however, for the latter the current guidelines may overlook important details. In the case of the most common laboratory species, the mouse, the range of frequencies they can hear is far higher than that of humans. The current guidelines briefly mention that ultrasonic (>20 kHz) frequencies can adversely affect mice, and that the acoustic environment should be checked, though no recommendations are provided relating to acceptable levels of ultrasonic noise. Methods: To investigate the ultrasonic environment in a large mouse breeding facility (the Mary Lyon Centre at MRC Harwell), we compared two systems, the Hottinger Bruel and Kjaer PULSE sound analyser, and an Avisoft Bioacoustics system. Potential noise sources were selected; we used the PULSE system to undertake real-time Fourier analysis of noise up to 100 kHz, and the Avisoft system to record noise up to 125 kHz for later analysis. The microphones from both systems were positioned consistently at the same distance from the source and environmental conditions were identical. In order to investigate our result further, a third system, the AudioMoth (Open Acoustic Devices), was also used for recording. We used DeepSqueak software for most of the recording analysis and, in some cases, we also undertook further spectral analysis using RX8 (iZotope, USA). Results: We found that both systems can detect a range of ultrasonic noise sources, and here discuss the benefits and limitations of each approach. Conclusions: We conclude that measuring the acoustic environment of animal facilities, including ultrasonic frequencies that may adversely affect the animals housed, will contribute to minimising disruption to animal welfare and perturbations in scientific research.
Collapse
Affiliation(s)
- Andrew Parker
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, UK
| | - Liane Hobson
- Mary Lyon Centre at MRC Harwell, Harwell Science Campus, Oxford, UK
| | - Rasneer Bains
- Mary Lyon Centre at MRC Harwell, Harwell Science Campus, Oxford, UK
| | - Sara Wells
- Mary Lyon Centre at MRC Harwell, Harwell Science Campus, Oxford, UK
| | - Michael Bowl
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, UK
- UCL Ear Institute, University College London, London, UK
| |
Collapse
|
11
|
Li Q, Li H, Yao X, Wang C, Liu H, Xu D, Yang C, Zhuang H, Xiao Y, Liu R, Shen S, Zhou S, Fu C, Wang Y, Teng G, Liu L. Stress Response and Hearing Loss Differentially Contribute to Dynamic Alterations in Hippocampal Neurogenesis and Microglial Reactivity in Mice Exposed to Acute Noise Exposure. Front Neurosci 2021; 15:749925. [PMID: 34955715 PMCID: PMC8692372 DOI: 10.3389/fnins.2021.749925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Noise-induced hearing loss (NIHL) is one of the most prevalent forms of acquired hearing loss, and it is associated with aberrant microglial status and reduced hippocampal neurogenesis; however, the nature of these associations is far from being elucidated. Beyond its direct effects on the auditory system, exposure to intense noise has previously been shown to acutely activate the stress response, which has increasingly been linked to both microglial activity and adult hippocampal neurogenesis in recent years. Given the pervasiveness of noise pollution in modern society and the important implications of either microglial activity or hippocampal neurogenesis for cognitive and emotional function, this study was designed to investigate how microglial status and hippocampal neurogenesis change over time following acoustic exposure and to analyze the possible roles of the noise exposure-induced stress response and hearing loss in these changes. To accomplish this, adult male C57BL/6J mice were randomly assigned to either a control or noise exposure (NE) group. Auditory function was assessed by measuring ABR thresholds at 20 days post noise exposure. The time-course profile of serum corticosterone levels, microglial status, and hippocampal neurogenesis during the 28 days following noise exposure were quantified by ELISA or immunofluorescence staining. Our results illustrated a permanent moderate-to-severe degree of hearing loss, an early but transient increase in serum corticosterone levels, and time-dependent dynamic alterations in microglial activation status and hippocampal neurogenesis, which both present an early but transient change and a late but enduring change. These findings provide evidence that both the stress response and hearing loss contribute to the dynamic alterations of microglia and hippocampal neurogenesis following noise exposure; moreover, noise-induced permanent hearing loss rather than noise-induced transient stress is more likely to be responsible for perpetuating the neurodegenerative process associated with many neurological diseases.
Collapse
Affiliation(s)
- Qian Li
- School of Life Science and Technology, Southeast University, Nanjing, China
| | - Hong Li
- School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xiuting Yao
- Medical College, Southeast University, Nanjing, China
| | - Conghui Wang
- Medical College, Southeast University, Nanjing, China
| | - Haiqing Liu
- School of Life Science and Technology, Southeast University, Nanjing, China
| | - Dan Xu
- School of Public Health, Southeast University, Nanjing, China
| | - Chenxi Yang
- Medical College, Southeast University, Nanjing, China
| | - Hong Zhuang
- Medical College, Southeast University, Nanjing, China
| | - Yu Xiao
- Medical College, Southeast University, Nanjing, China
| | - Rui Liu
- Medical College, Southeast University, Nanjing, China
| | - Sinuo Shen
- Medical College, Southeast University, Nanjing, China
| | - Shaoyang Zhou
- Medical College, Southeast University, Nanjing, China
| | - Chenge Fu
- Medical College, Southeast University, Nanjing, China
| | - Yifan Wang
- Medical College, Southeast University, Nanjing, China
| | - Gaojun Teng
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Medical School, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lijie Liu
- Medical College, Southeast University, Nanjing, China
| |
Collapse
|
12
|
Paciello F, Rinaudo M, Longo V, Cocco S, Conforto G, Pisani A, Podda MV, Fetoni AR, Paludetti G, Grassi C. Auditory sensory deprivation induced by noise exposure exacerbates cognitive decline in a mouse model of Alzheimer's disease. eLife 2021; 10:70908. [PMID: 34699347 PMCID: PMC8547960 DOI: 10.7554/elife.70908] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022] Open
Abstract
Although association between hearing impairment and dementia has been widely documented by epidemiological studies, the role of auditory sensory deprivation in cognitive decline remains to be fully understood. To address this issue we investigated the impact of hearing loss on the onset and time-course of cognitive decline in an animal model of Alzheimer's disease (AD), that is the 3×Tg-AD mice and the underlying mechanisms. We found that hearing loss induced by noise exposure in the 3×Tg-AD mice before the phenotype is manifested caused persistent synaptic and morphological alterations in the auditory cortex. This was associated with earlier hippocampal dysfunction, increased tau phosphorylation, neuroinflammation, and redox imbalance, along with anticipated memory deficits compared to the expected time-course of the neurodegenerative phenotype. Our data suggest that a mouse model of AD is more vulnerable to central damage induced by hearing loss and shows reduced ability to counteract noise-induced detrimental effects, which accelerates the neurodegenerative disease onset.
Collapse
Affiliation(s)
- Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Longo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Cocco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Conforto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Vittoria Podda
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gaetano Paludetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
13
|
Farazi N, Mahmoudi J, Sadigh-Eteghad S, Farajdokht F, Rasta SH. Synergistic effects of combined therapy with transcranial photobiomodulation and enriched environment on depressive- and anxiety-like behaviors in a mice model of noise stress. Lasers Med Sci 2021; 37:1181-1191. [PMID: 34432186 DOI: 10.1007/s10103-021-03370-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/03/2021] [Indexed: 12/11/2022]
Abstract
The development of anxiety and depression due to chronic exposure to noise stress has remained as an unsolved health problem so far. Despite the studies suggesting the neuroenhancement effects of transcranial photobiomodulation (tPBM) and housing in an enriched environment (EE), the combined effects of these treatments have not been elucidated yet. Also, there is no available data on the relationship between the application of tPBM and hippocampal brain-derived neurotrophic factor (BDNF) expression in animal models of stress. The present study aims to investigate the application of the tPBM and EE (alone or in combination) on depressive- and anxiety-like behaviors in a mice model of noise stress. Mice were divided into five groups: control, noise, noise + EE, noise + tPBM, and noise + EE + tPBM. Except for the control group, other groups were subjected to 110 dB SPL white noise for 4 h/day for 14 consecutive days and received their respective treatments. Forced Swimming Test (FST) was used to evaluate depressive-like behaviors. Elevated Plus Maze (EPM) and Open Field Test (OFT) were used to evaluate anxiety-like behaviors. BDNF, tyrosine receptor kinase B (TrkB), and cAMP response element-binding (CREB) protein levels in the hippocampus were determined by the Western blot method, and also serum corticosterone levels were assessed using an ELISA kit. Exposure to noise stress significantly elevated serum corticosterone level; downregulated hippocampal BDNF, TrkB, and CREB protein expressions; and resulted in depressive- and anxiety-like behaviors. While, the application of tPBM (810 nm wavelength, 8 J/cm2 fluence, 10 Hz pulsed wave mode), housing in EE, and their combination lowered corticosterone levels, upregulated the BDNF/TrkB/CREB signaling pathway in the hippocampus, and improved behavioral outcomes in noise stress subjected mice. Our finding revealed the improving effects of tPBM and EE on depressive and anxiety-like behaviors induced by noise stress, possibly by augmenting the BDNF/TrkB/CREB signaling pathway.
Collapse
Affiliation(s)
- Narmin Farazi
- Department of Medical Physics, Tabriz University of Medical Sciences, 51666-14766, Tabriz, Iran.,Neurosciences Research Center, Tabriz University of Medical Sciences, 51666-14756, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, 51666-14756, Tabriz, Iran.
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, 51666-14756, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, 51666-14756, Tabriz, Iran
| | - Seyed Hossein Rasta
- Department of Medical Physics, Tabriz University of Medical Sciences, 51666-14766, Tabriz, Iran. .,Department of Medical Bioengineering, Tabriz University of Medical Sciences, Tabriz, Iran. .,School of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
14
|
de Deus JL, Amorim MR, Ribeiro AB, Barcellos-Filho PCG, Ceballos CC, Branco LGS, Cunha AOS, Leão RM. Loss of Brain-Derived Neurotrophic Factor Mediates Inhibition of Hippocampal Long-Term Potentiation by High-Intensity Sound. Cell Mol Neurobiol 2021; 41:751-763. [PMID: 32445041 PMCID: PMC11448697 DOI: 10.1007/s10571-020-00881-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/16/2020] [Indexed: 01/15/2023]
Abstract
Exposure to noise produces cognitive and emotional disorders, and recent studies have shown that auditory stimulation or deprivation affects hippocampal function. Previously, we showed that exposure to high-intensity sound (110 dB, 1 min) strongly inhibits Schaffer-CA1 long-term potentiation (LTP). Here we investigated possible mechanisms involved in this effect. We found that exposure to 110 dB sound activates c-fos expression in hippocampal CA1 and CA3 neurons. Although sound stimulation did not affect glutamatergic or GABAergic neurotransmission in CA1, it did depress the level of brain-derived neurotrophic factor (BDNF), which is involved in promoting hippocampal synaptic plasticity. Moreover, perfusion of slices with BDNF rescued LTP in animals exposed to sound stimulation, whereas BDNF did not affect LTP in sham-stimulated rats. Furthermore, LM22A4, a TrkB receptor agonist, also rescued LTP from sound-stimulated animals. Our results indicate that depression of hippocampal BDNF mediates the inhibition of LTP produced by high-intensity sound stimulation.
Collapse
Affiliation(s)
- Júnia L de Deus
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-904, Brazil
| | - Mateus R Amorim
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-904, Brazil
| | - Aline B Ribeiro
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Procópio C G Barcellos-Filho
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - César C Ceballos
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luiz Guilherme S Branco
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-904, Brazil
| | - Alexandra O S Cunha
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Ricardo M Leão
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
15
|
|
16
|
Zhuang H, Yang J, Huang Z, Liu H, Li X, Zhang H, Wang J, Yu S, Liu K, Liu R, Bi M, Wang J, Salvi RJ, Hu B, Teng G, Liu L. Accelerated age-related decline in hippocampal neurogenesis in mice with noise-induced hearing loss is associated with hippocampal microglial degeneration. Aging (Albany NY) 2020; 12:19493-19519. [PMID: 33041264 PMCID: PMC7732316 DOI: 10.18632/aging.103898] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/23/2020] [Indexed: 01/24/2023]
Abstract
Large-scale epidemiological surveys suggest that hearing loss (HL) is a significant risk factor for dementia. We previously showed that noise-induced HL (NIHL) impairs hippocampal cognitive function and decreases hippocampal neurogenesis and neuronal complexity, suggesting a causal role of HL in dementia. To further investigate the influence of acquired peripheral HL on hippocampal neurogenesis with the aging process as well as the underlying mechanism, we produced NIHL in male CBA/J mice and assessed hippocampal neurogenesis and microglial morphology in the auditory brain and hippocampus at 4 days post-noise exposure (DPN) or 1, 3, 6, or 12 months post-noise exposure (MPN) by immunofluorescence labeling. We found that the age-related decline in hippocampal neurogenesis was accelerated in mice with NIHL. Furthermore, in mice with NIHL, prolonged microglial activation occurred from 1 MPN to 12 MPN across multiple auditory nuclei, while aggravated microglial deterioration occurred in the hippocampus and correlated with the age-related decline in hippocampal neurogenesis. These results suggest that acquired peripheral HL accelerates the age-related decline in hippocampal neurogenesis and that hippocampal microglial degeneration may contribute to the development of neurodegeneration following acquired peripheral HL.
Collapse
Affiliation(s)
- Hong Zhuang
- Department of Physiology, Medical College, Southeast University, Nanjing 210009, China
| | - Jing Yang
- Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Zhihui Huang
- Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Haiqing Liu
- Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Xiaobo Li
- Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Hongyu Zhang
- Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Jiadong Wang
- Medical College, Southeast University, Nanjing 210009, China
| | - Shen Yu
- Medical College, Southeast University, Nanjing 210009, China
| | - Kefei Liu
- Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Rui Liu
- Medical College, Southeast University, Nanjing 210009, China
| | - Mingze Bi
- Medical College, Southeast University, Nanjing 210009, China
| | - Jian Wang
- School of Human Communication Disorder, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Richard J. Salvi
- Center for Hearing and Deafness, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Bohua Hu
- Center for Hearing and Deafness, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Gaojun Teng
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Department of Physiology, Medical College, Southeast University, Nanjing 210009, China
| |
Collapse
|
17
|
Haider S, Sajid I, Batool Z, Madiha S, Sadir S, Kamil N, Liaquat L, Ahmad S, Tabassum S, Khaliq S. Supplementation of Taurine Insulates Against Oxidative Stress, Confers Neuroprotection and Attenuates Memory Impairment in Noise Stress Exposed Male Wistar Rats. Neurochem Res 2020; 45:2762-2774. [PMID: 32918662 DOI: 10.1007/s11064-020-03127-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/07/2020] [Accepted: 09/05/2020] [Indexed: 12/30/2022]
Abstract
Noise has always been an important environmental factor that induces health problems in the general population. Due to ever increasing noise pollution, humans are facing multiple auditory and non-auditory problems including neuropsychiatric disorders. In modern day life it is impossible to avoid noise due to the rapid industrialization of society. Continuous exposure to noise stress creates a disturbance in brain function which may lead to memory disorder. Therefore, it is necessary to find preventive measures to reduce the deleterious effects of noise exposure. Supplementation of taurine, a semi essential amino acid, is reported to alleviate psychiatric disorders. In this study noise-exposed (100 db; 3 h daily for 15 days) rats were supplemented with taurine at a dose of 100 mg/kg for 15 days. Spatial and recognition memory was assessed using the Morris water maze and novel object recognition task, respectively. Results of this study showed a reversal of noise-induced memory impairment in rats. The derangements of catecholaminergic and serotonergic levels in the hippocampus and altered brain antioxidant enzyme activity due to noise exposure were also restored by taurine administration. This study highlights the importance of taurine supplementation to mitigate noise-induced impaired memory via normalizing the neurochemical functions and reducing oxidative stress in rat brain.
Collapse
Affiliation(s)
- Saida Haider
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Irfan Sajid
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
- Department of Biochemistry, Federal Urdu University of Arts, Science and Technology, Karachi, Pakistan
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and BiologicalSciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Syeda Madiha
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Sadia Sadir
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Noor Kamil
- Department of Basic Medical Sciences, Faculty of Pharmacy, Barrett Hodgson University, Karachi, Pakistan
| | - Laraib Liaquat
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Saara Ahmad
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Saiqa Tabassum
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
- Department of Biochemistry, Shaheed Zuifiqar Ali Bhutto Institute of Science and Technology, Karachi, Pakistan
| | - Saima Khaliq
- Department of Biochemistry, Federal Urdu University of Arts, Science and Technology, Karachi, Pakistan
| |
Collapse
|
18
|
Neuroanatomical changes of the medial prefrontal cortex of male pups of Wistar rat after prenatal and postnatal noise stress. Acta Histochem 2020; 122:151589. [PMID: 32778245 DOI: 10.1016/j.acthis.2020.151589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022]
Abstract
Recent evidences showed that, noise stress causes abnormal changes in structure and function of central nervous system (CNS). The Current study was conducted to evaluate some stereological parameters of the medial prefrontal cortex (mPFC) of male pups of Wistar rat after prenatal and early postnatal noise stress. 18 pregnant Wistar rats were randomly divided into prenatal noise-exposed (NE) group, postnatal NE group, and controls. Male pups of NE groups were exposed to noise 100 dB at the frequency ranges of 500-8000 Hz, 4 h per day from gestational day one (GD1) to GD21 for the prenatal NE group, and from postnatal day one (PND1) to PND21 in the postnatal NE group. The Control group animals were maintained under standard condition without noise stimulation. Corticosterone level in plasma was measured using ELISA technique. Changes of the neurons and non-neurons cells number and volume of the mPFC were evaluated by stereological analysis. Tunnel assay was also used for detection of apoptotic cells. Increase in plasma corticosterone level, decrease in the number of neurons, and increase in the apoptotic cells number were observed in both NE groups. Decrease in volume of mPFC and also in non-neurons cells number was observed in the prenatal NE group. An increase in the non-neurons number was seen in the postnatal NE group. Data of the current comparative study showed that, noise stress during prenatal and early postnatal periods can induce the abnormal alteration in some stereological parameters of mPFC in male pups of Wistar rat. These negative alterations were more remarkable after prenatal noise stress.
Collapse
|
19
|
Akefe IO, Ayo JO, Sinkalu VO. Kaempferol and zinc gluconate mitigate neurobehavioral deficits and oxidative stress induced by noise exposure in Wistar rats. PLoS One 2020; 15:e0236251. [PMID: 32692754 PMCID: PMC7373279 DOI: 10.1371/journal.pone.0236251] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
This study investigated the effects of kaempferol and zinc gluconate on neurobehavioural and oxidative stress changes in Wistar rats exposed to noise. Thirty (30) rats were randomly divided into five groups: Groups I and II were administered with deionized water (DW); Group III, kaempferol (K); Group IV, zinc gluconate (Zn); Group V, kaempferol + zinc gluconate. Groups II, III, IV, and V were subjected to noise stress (N) induced by exposing rats to 100 dB (4 h/day) for 15 days, from day 33 to day 48 after starting the drug treatments. Neuromuscular coordination, motor coordination, motor strength, sensorimotor reflex, and learning and memory, were evaluated using standard laboratory methods. Levels of nitric oxide (NO), malondialdehyde (MDA) and activities of glutathione peroxidase (GPx), catalase and superoxide dismutase (SOD) were evaluated in the hippocampus. Exposure of rats to noise, induced significant neurobehavioural deficits and oxidative stress while the combined administration of kaempferol and zinc gluconate significantly (P < 0.05) improved open-field performance, motor coordination, motor strength, sensorimotor reflex, and learning and memory. Co-administration of kaempferol and zinc gluconate ameliorated noise-induced oxidative stress as demonstrated by the significantly increased activities of GPx, catalase, and SOD, and decreased levels of NO and MDA (P < 0.05 and P < 0.01 respectively), compared to the DW + N group. Our results suggest that oxidative stress, evidenced by increased NO and MDA concentration and decreased activities of GPx, catalase and SOD, were involved in the molecular mechanism underlying neurobehavioural impairment in Wistar rats, exposed to noise stress. Single treatment of kaempferol exerted a more potent mitigative effect than zinc gluconate, while their combination produced an improved outcome.
Collapse
Affiliation(s)
- Isaac Oluwatobi Akefe
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Jos, Jos, Nigeria
- Department of Physiology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Nigeria
- * E-mail:
| | - Joseph Olusegun Ayo
- Department of Physiology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Nigeria
| | - Victor Olusegun Sinkalu
- Department of Physiology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
20
|
Luchese C, Vogt AG, Pinz MP, Dos Reis AS, Gomes CB, Alves D, Wilhelm EA. Amnesia-ameliorative effect of a quinoline derivative through regulation of oxidative/cholinergic systems and Na +/K +-ATPase activity in mice. Metab Brain Dis 2020; 35:589-600. [PMID: 32048104 DOI: 10.1007/s11011-020-00535-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
The present study evaluated the anti-amnesic activity of 1-(7-chloroquinolin-4-yl)-5-methyl-N-phenyl-1H-1,2,3-triazole-4-carboxamide (QTCA-1) against scopolamine (SCO)-induced amnesia in mice. It was evaluated cholinergic dysfunction, oxidative stress and Na+/K+-ATPase activity in cerebral cortex and hippocampus of mice. Male Swiss mice were treated with QTCA-1 (10 mg/kg, intragastrically (i.g.), daily) for nine days. Thirty minutes after the treatment with compound, the animals received a injection of SCO (0.4 mg/kg, intraperitoneally (i.p.)). Mice were submitted to the behavioral tasks 30 min after injection of SCO (Barnes maze, open-field, object recognition and location, and step-down inhibitory avoidance tasks) during nine days. In day 9, cerebral cortex and hippocampus of mice were removed to determine the thiobarbituric acid reactive species (TBARS) levels, and catalase (CAT), Na+/K+-ATPase and acetylcholinesterase (AChE) activities. SCO caused amnesia in mice for changing in step-down inhibitory avoidance, Barnes maze, and object recognition and object location tasks. QTCA-1 treatment attenuated the behavioral changes caused by SCO. Moreover, SCO increased AChE and CAT activities, decreased Na+/K+-ATPase activity and increased TBARS levels in the cerebral structures of mice. QTCA-1 protected against these brain changes. In conclusion, QTCA-1 had anti-amnesic action in the experimental model used in the present study, through the anticholinesterase effect, modulation of Na+/K+-ATPase activity and antioxidant action.
Collapse
Affiliation(s)
- Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brazil.
| | - Ane G Vogt
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brazil
| | - Mikaela P Pinz
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brazil
| | - Angélica S Dos Reis
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brazil
| | - Carolina B Gomes
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa - (LASOL), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), P.O. Box 354, Pelotas, RS, 96010-900, Brazil
| | - Diego Alves
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa - (LASOL), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), P.O. Box 354, Pelotas, RS, 96010-900, Brazil
| | - Ethel A Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brazil.
| |
Collapse
|
21
|
Maghami S, Zardooz H, Khodagholi F, Binayi F, Ranjbar Saber R, Hedayati M, Sahraei H, Ansari MA. Maternal separation blunted spatial memory formation independent of peripheral and hippocampal insulin content in young adult male rats. PLoS One 2018; 13:e0204731. [PMID: 30332425 PMCID: PMC6192583 DOI: 10.1371/journal.pone.0204731] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/13/2018] [Indexed: 01/22/2023] Open
Abstract
This study explores the effects of maternal separation as a chronic early life stress (ELS) on pancreatic islets insulin content and secretion, and their potential relationship with the hippocampus insulin content and spatial memory in young adulthood. Male rat offspring were divided into two groups: stress (STR) and non-stress (non-STR) groups. The animals of the STR group were separated from their mothers during postnatal days (PND) 1 to 21. During the weaning time, that is, PND-0 to PND-21, the body weight and length of the pups were measured. Blood samples were collected on PND-1, 21, 29 and 34 and during young adulthood (53±2 days) to determine plasma corticosterone and insulin levels. The young adult animals were also tested for spatial memory. One day after the memory test, the animals were decapitated and their pancreases were removed to measure the islets insulin content and secretion. Finally, the animals' hippocampi were isolated to determine their insulin content and insulin receptor protein amounts. During the period of weaning, the body weight and length of pups belonging to the STR group were significantly lower as compared to those in the non-STR group. Maternal separation did not change the plasma levels of insulin but increased plasma corticosterone levels from PND-21 to young adulthood and also reduced the islets insulin content but did not affect insulin secretion and the hippocampus insulin content and insulin receptor protein amount. Although, at the end of the memory tests, rats of the STR group reached the escape box at almost the same time and distance and with the same errors as rats of the non-STR group, the distance traveled to reach the escape box showed a steep reduction in the non-STR group as compared to the STR group after the first trial. Moreover, as compared to the STR group, the non-STR group showed an increasing trend for direct strategy to find the escape box. The islets insulin content and secretion, and the plasma insulin concentration were not significantly correlated with the hippocampus insulin content. From the results of the present study, it appears that the main behavioral effect of the maternal separation stress in the spatial memory task was to impair the strategy used by the animals to reach the escape box. This may indicate that maternal separation stress affects brain regions other than the hippocampus. Moreover, due to the reduction of the body weight and length of offspring belonging to the STR group, it should be further considered that both maternal separation and early life malnutrition are directly (and mechanistically) linked to cognitive alterations later in life in ways that are not dependent on peripheral and hippocampal insulin content.
Collapse
Affiliation(s)
- Soheila Maghami
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- * E-mail: ,
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Ranjbar Saber
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Ansari
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
22
|
Liu L, Xuan C, Shen P, He T, Chang Y, Shi L, Tao S, Yu Z, Brown RE, Wang J. Hippocampal Mechanisms Underlying Impairment in Spatial Learning Long After Establishment of Noise-Induced Hearing Loss in CBA Mice. Front Syst Neurosci 2018; 12:35. [PMID: 30087600 PMCID: PMC6066960 DOI: 10.3389/fnsys.2018.00035] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/03/2018] [Indexed: 12/20/2022] Open
Abstract
Sensorineural hearing loss (SNHL) has been demonstrated in many clinical reports as a risk factor that promotes the development of cognitive impairment. However, the underlying neurological mechanisms are not clear. Noise exposure is one of the most common causes of SNHL. Although noise exposure causes relatively less damage to general health as compared with other methods for creating hearing loss (such as ototoxicity), it does impair cognitive function. Many studies have shown that the noise-induced cognitive impairment occur via the oxidative stress induced by the noise. In those studies, the effects of the noise-induced hearing loss induced (NIHL) were not addressed. Previously, we have demonstrated in the CBA/CaJ mouse model that oxidative stress was transient after a brief noise exposure, but the NIHL was permanent. In addition, NIHL was followed by a declined cognitive function and decreased hippocampal neurogenesis that were developed long after the oxidative stress disappeared. Therefore, NIHL can cause cognitive impairment independent of its stress effect and can serve as a model to investigate the relationship between hearing loss and the development of cognitive impairment. In the present study, we further demonstrated that the oxidative stress produced by the brief noise exposure did not damage the stem cell bank of hippocampus that was evaluated shortly after the noise exposure. In addition to the reduction in the rate of cell proliferation in hippocampus that was found previously, we found that the NIHL significantly reduced the promoting effect of learning activity on various stages of hippocampal neurogenesis, accompanied by the reduction in learning-induced expression of immediate early genes (IEGs) in hippocampus. Since the MWM-tested spatial function does not directly require auditory input, the results provide evidence for the maintenance role of auditory input on the cognitive function; the reduction of IEG expression that is required in memory-formation may be the initial step in blocking the effect of learning activity on neurogenesis in subjects with NIHL.
Collapse
Affiliation(s)
- Lijie Liu
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| | - Chuanying Xuan
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Pei Shen
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Tingting He
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Ying Chang
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Lijuan Shi
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| | - Shan Tao
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| | - Zhiping Yu
- School of Communication Science and Disorders, Dalhousie University, Halifax, NS, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Jian Wang
- Department of Physiology, Medical College, Southeast University, Nanjing, China.,School of Communication Science and Disorders, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
23
|
Scopolamine-induced greater alterations in neurochemical profile and increased oxidative stress demonstrated a better model of dementia: A comparative study. Brain Res Bull 2016; 127:234-247. [DOI: 10.1016/j.brainresbull.2016.10.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/05/2016] [Indexed: 02/08/2023]
|